US20180148763A1 - Method for quantifying monoclonal antibody - Google Patents

Method for quantifying monoclonal antibody Download PDF

Info

Publication number
US20180148763A1
US20180148763A1 US15/578,374 US201515578374A US2018148763A1 US 20180148763 A1 US20180148763 A1 US 20180148763A1 US 201515578374 A US201515578374 A US 201515578374A US 2018148763 A1 US2018148763 A1 US 2018148763A1
Authority
US
United States
Prior art keywords
protease
monoclonal antibody
antibody
fine particles
trypsin
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US15/578,374
Other languages
English (en)
Inventor
Takashi Shimada
Noriko IWAMOTO
Akinobu Hamada
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
NATIONAL CANCER CENTER
Shimadzu Corp
National Cancer Center Japan
Original Assignee
Shimadzu Corp
National Cancer Center Japan
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Shimadzu Corp, National Cancer Center Japan filed Critical Shimadzu Corp
Assigned to SHIMADZU CORPORATION, NATIONAL CANCER CENTER reassignment SHIMADZU CORPORATION ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: IWAMOTO, NORIKO, SHIMADA, TAKASHI, HAMADA, AKINOBU
Publication of US20180148763A1 publication Critical patent/US20180148763A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N27/00Investigating or analysing materials by the use of electric, electrochemical, or magnetic means
    • G01N27/62Investigating or analysing materials by the use of electric, electrochemical, or magnetic means by investigating the ionisation of gases, e.g. aerosols; by investigating electric discharges, e.g. emission of cathode
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/34Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving hydrolase
    • C12Q1/37Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving hydrolase involving peptidase or proteinase
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y304/00Hydrolases acting on peptide bonds, i.e. peptidases (3.4)
    • C12Y304/21Serine endopeptidases (3.4.21)
    • C12Y304/21001Chymotrypsin (3.4.21.1)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y304/00Hydrolases acting on peptide bonds, i.e. peptidases (3.4)
    • C12Y304/21Serine endopeptidases (3.4.21)
    • C12Y304/21004Trypsin (3.4.21.4)
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N30/00Investigating or analysing materials by separation into components using adsorption, absorption or similar phenomena or using ion-exchange, e.g. chromatography or field flow fractionation
    • G01N30/02Column chromatography
    • G01N30/62Detectors specially adapted therefor
    • G01N30/72Mass spectrometers
    • G01N30/7233Mass spectrometers interfaced to liquid or supercritical fluid chromatograph
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/94Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving narcotics or drugs or pharmaceuticals, neurotransmitters or associated receptors
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • G01N2333/705Assays involving receptors, cell surface antigens or cell surface determinants
    • G01N2333/715Assays involving receptors, cell surface antigens or cell surface determinants for cytokines; for lymphokines; for interferons
    • G01N2333/7158Assays involving receptors, cell surface antigens or cell surface determinants for cytokines; for lymphokines; for interferons for chemokines

Definitions

  • the present invention relates to a method for quantifying a monoclonal antibody using mass spectrometry, and more specifically relates to a method that includes a step of selectively digesting a peptide fragment that includes a specific sequence of a monoclonal antibody, and a step of detecting a resulting peptide fragment using mass spectrometry, and in which a specific enzyme for selective digestion is used.
  • concentration monitoring therapeutic drug monitoring (TDM) has attracted attention for development and administration of drugs that have less side effects and exhibit high medicinal effects.
  • concentration monitoring whether or not an administered drug is in a proper amount and whether or not the drug has reached a lesion site can be confirmed, and effectiveness of the drug can be evaluated and dosage can be appropriately adjusted. Further, it is important that the effectiveness of a molecularly targeted drug, which has become a mainstream in areas such as cancer and autoimmune diseases, can be quickly judged by a doctor himself or herself based on whether or not the drug accumulates at a lesion site and exerts its medicinal effect.
  • Antibody drugs are attracting attention as molecular target drugs.
  • Antibody drugs specifically bind to cancer and other lesion antigens, target molecules, growth factors, and the like, and thus have attracted attentions as drugs that have less side effects and exhibit high medicinal effects, and clinical studies have been carried out with many antibodies.
  • Antibodies exhibit extremely high molecular specificity due to their nature.
  • cases are often seen such as a case where antibodies do not exert their medicinal effects merely due to differences in lesion sites, and a case where antibodies are not effective for metastatic lesions.
  • drug prices and it is argued that it is important to conduct optimized medical care by proper use of antibody drugs.
  • ELISA Enzyme-Linked ImmunoSorbent Assay
  • a group including the present inventors has found a method that allows an optimal peptide fragment to be obtained for specific detection of a protein such as an antibody using mass spectrometry by selective protease digestion of the protein.
  • This method realizes regioselective protease digestion of a monoclonal antibody by immobilizing both a protein such as an antibody as a substrate and a protease enzyme on solid phases (Patent Document 1 and Non-Patent Document 1).
  • Patent Document 1 and Non-Patent Document 1 is an innovative method that allows a selective protease digestion of a monoclonal antibody to be performed using a solid phase-solid phase reaction. More specifically, for example, a C-terminal side of an antibody is immobilized onto a Protein G or a Protein A resin having a pore diameter of about 100 nm, and a variable region of the antibody is always oriented to a solution side. Next, a protease is immobilized on surfaces of fine particles having a particle size of about 200 nm. By limiting contact of a protease with the antibody, it is possible to form a reaction field in which a variable region selective antibody decomposition reaction is performed.
  • the present inventors have found that, in order to detect an antibody using limiting protease digestion, it is most effective to detect a protease digestion fragment contained in particular in a CDR2 region in a variable region of a monoclonal antibody.
  • a protease having less self-digestion and high cleavage sequence selectivity when a commercially available protease is used, a protease of a mass spectrometry grade or of a sequencing (sequence) grade is used.
  • a native trypsin derived from a living body generates a pseudo trypsin exhibiting chymotrypsin-like activity by self-digestion and thus is known to have low cleavage site specificity. Therefore, a trypsin of a mass spectrometry grade having improved resistance to self-digestion by subjecting a lysine residue of the trypsin to reductive methylation is commercially available.
  • the present inventors believe that the above result suggests that there may be interference due to presence of an endogenous antibody to the obtained peptide fragment, and have further studied for a purpose of obtaining a peptide fragment without such interference.
  • an enzyme with lower specificity than a conventional structural analysis enzyme it was surprisingly found that, by using an enzyme with lower specificity than a conventional structural analysis enzyme, the number of cutting sites is increased, peptide fragments of different sequences are generated, and, consequently, optimal peptides for quantitative detection can be obtained, and thus, the present invention is accomplished.
  • the present invention includes the following aspects.
  • a method for quantifying a monoclonal antibody including: a step of performing selective protease digestion of a monoclonal antibody as a measurement target by bringing a porous body in which the monoclonal antibody is immobilized in pores into contact with fine particles onto which a specific protease is immobilized, the fine particles having an average particle size larger than an average pore diameter of the porous body; and a step of detecting a peptide fragment that is obtained from the digestion and contains an amino acid derived from a CDR2 region of a heavy chain or a light chain of the monoclonal antibody, wherein, as the protease, a protease having trypsin activity and chymotrypsin activity is used.
  • the protease having trypsin activity and chymotrypsin activity is a trypsin that has not been subjected to a reductive methylation treatment.
  • the monoclonal antibody is mogamulizumab.
  • the peptide fragment is a peptide having an amino acid sequence expressed in SEQ ID NO: 10.
  • a kit for quantitative detection of a monoclonal antibody using liquid chromatography-mass spectrometry including: a porous body for immobilizing a monoclonal antibody as a measurement target; fine particles onto which a protease having trypsin activity and chymotrypsin activity is immobilized; a reaction container for selectively digesting the monoclonal antibody by bringing the porous body into contact with the fine particles; and a buffer solution that is introduced into the reaction container together with the fine particles and the porous body and is for causing a digestion reaction by the protease to occur.
  • LC-MS liquid chromatography-mass spectrometry
  • the protease having trypsin activity and chymotrypsin activity is a trypsin that has not been subjected to a reductive methylation treatment.
  • the measurement target is mogamulizumab
  • an internal standard peptide is a peptide having an amino acid sequence expressed in SEQ ID NO: 10.
  • a computer readable recording medium that is for use in the method described in any one of the aspects (1)-(4) and in which data for executing mass spectrometry is recorded, wherein the data includes data of a parent ion, a fragment ion, and a voltage applied to electrodes of a mass spectrometer, with respect to a peptide obtained by protease digestion of the monoclonal antibody.
  • LC-MS liquid chromatography-mass spectrometry
  • the method of the present invention uses a limiting protease reaction field, it is possible to selectively decompose and collect a limited region of an antibody, in particular, a CDR2 region.
  • the CDR2 region is an exposed part closest to a surface of the antibody and thus is preferentially decomposed. Utilizing this fact, detection of a monoclonal antibody using mass spectrometry can be more efficiently performed.
  • concentration of the antibody drug can be directly measured from a biological sample.
  • quantification of individual antibodies using mass spectrometry can be reliably performed. Further, when standard analysis conditions with respect to individual antibodies are provided, a doctor himself or herself can be involved in analysis of a specimen at a clinical site, and effectiveness of an administered antibody can be promptly evaluated.
  • FIG. 1 illustrates amino acid sequences (SEQ ID NO: 1 and 2) of a heavy chain and a light chain of mogamulizumab, and sequences of Trypsin Gold cleavage fragments used for quantification and positions of the sequences in the amino acid sequence of the heavy chain or the light chain.
  • FIG. 2 illustrates MRM chromatograms of Trypsin Gold cleavage fragments (SEQ ID NO: 3-9) of mogamulizumab.
  • An upper figure illustrates analysis results of mogamulizumab raw powder, and a lower figure illustrates analysis results of a sample obtained by spiking mogamulizumab into a plasma.
  • a vertical axis indicates a peak intensity and a horizontal axis indicates a retention time. Fragments derived from the same peptide are detected at the same retention time and a difference in production efficiency of a fragment ion is indicated by a peak height.
  • FIG. 3 illustrates calibration curves prepared using Trypsin Gold cleavage fragments (SEQ ID NO: 3-9) of mogamulizumab spiked into a plasma.
  • FIG. 4 illustrates sequences of Trypsin TPCK-Treated cleavage fragments used in quantification of mogamulizumab, and positions of the sequences in amino acid sequences of a heavy chain and a light chain.
  • FIG. 5 illustrates MRM chromatograms of Trypsin TPCK-Treated cleavage fragments (SEQ ID NO: 10-14) of mogamulizumab.
  • An upper figure illustrates analysis results of mogamulizumab raw powder, and a lower figure illustrates analysis results of a sample obtained by spiking mogamulizumab into a plasma.
  • a vertical axis indicates a peak intensity and a horizontal axis indicates a retention time. Fragments derived from the same peptide are detected at the same retention time and a difference in production efficiency of a fragment ion is indicated by a peak height.
  • FIG. 6 illustrates calibration curves prepared using Trypsin TPCK-Treated cleavage fragments (SEQ ID NO: 10-14) of mogamulizumab spiked into a plasma.
  • FIG. 7 illustrates structure identification results of mogamulizumab target peptide SYYPDSVK (SEQ ID NO: 10) in precision mass spectrometry.
  • the present invention relates to a method for quantifying a monoclonal antibody.
  • the method includes: a step of performing selective protease digestion of a monoclonal antibody as a measurement target by bringing a porous body in which the monoclonal antibody is immobilized in pores into contact with fine particles onto which a specific protease is immobilized, the fine particles having an average particle size larger than an average pore diameter of the porous body; and a step of detecting a peptide fragment that is obtained from the digestion and contains an amino acid derived from a CDR2 region of a heavy chain or a light chain of the monoclonal antibody.
  • the protease a protease having trypsin activity and chymotrypsin activity is used.
  • the method of the present invention is intended to reduce population to be analyzed while maintaining specificity of a measurement target.
  • the protease not a trypsin that is conventionally regarded as optimal for mass spectrometry, but a protease having trypsin activity and chymotrypsin activity is used. Thereby, a peptide fragment suitable for detection can be obtained.
  • CDR2 region
  • CDR2 region
  • FR framework region
  • a person skilled in the art can determine an amino acid sequence containing an amino acid derived from a CDR2 region for efficiently detecting specificity of a target antibody, and predict and select a peptide fragment as a detection target, by confirming multiple alignments of an amino acid sequence between a target antibody and another antibody and cross reactivity between the target antibody and an endogenous antibody.
  • amino acid sequence containing an amino acid derived from a CDR2 region means an amino acid sequence containing at least one amino acid derived from a CDR2 region specific to a target antibody, that is, an amino acid sequence containing a portion of the CDR2 region.
  • the specificity can be reliably detected.
  • amino acid sequence containing an amino acid derived from a CDR2 region is not intended to mean an arbitrary amino acid sequence containing “an amino acid derived from a CDR2 region”, but is intended to mean that the amino acid sequence as a whole forms a continuous sequence of a portion of an amino acid sequence of a target antibody.
  • a molecular weight of a peptide suitable for analysis is about 1000-3000 Da.
  • a measurement target in the method of the present invention is a monoclonal antibody.
  • a heavy chain and a light chain of a monoclonal antibody are each formed of a constant region and a variable region.
  • the constant regions each have an amino acid sequence that is common to most of antibodies derived from the same specie.
  • CDRs complementarity determining regions
  • CDR1, CDR2, CDR3 complementarity determining regions
  • the CDR regions involved in specific binding with an antigen are positioned substantially outside of an antibody molecule.
  • the CDR2 region is positioned on an outermost side ( FIG. 8 ), and is optimal as a target of the limiting protease digestion of the present invention.
  • an antibody that is clinically used an entire amino acid sequence thereof and sequences of the CDRs are disclosed. Further, a person skilled in the art can identify the CDR regions based on amino acid sequence information of the antibody.
  • a monoclonal antibody that can be a measurement target in the method of the present invention is not limited.
  • examples of the monoclonal antibody include: human antibodies such as mogamulizumab, panitumumab, offatumumab, golimumab, and ipilimumab; humanized antibodies such as tocilizumab, omalizumab, mepolizumab, gemtuzumab, palivizumab, ranibizumab, certolizumab, ocrelizumab, mogamulizumab, and eculizumab; chimeric antibodies such as cetuximab, infliximab, and basiliximab; and the like.
  • a monoclonal antibody has a molecular diameter of about 14.5 nm.
  • a CDR2 region of a monoclonal antibody can be regioselectively protease-digested, and identification and quantitative detection of the antibody can be performed by detecting a resulting peptide fragment of the CDR2 region using mass spectrometry.
  • the method of the present invention is applicable regardless of a type of an antibody, and thus, is not limited to the above exemplified antibodies, and is also applicable to newly developed monoclonal antibodies and the like.
  • a porous body used in the method of the present invention is not particularly limited in material as long as the porous body has a large number of pores, and activated carbon, a porous membrane, porous resin beads, metal particles, and the like, can be used. Among these, those capable of site-specifically binding an antibody are preferable.
  • the pores are not limited in shape. Further, as in a case of a porous membrane, a porous body having pores formed penetrating the porous body can also be used.
  • a size of a pore of a porous body is not particularly limited, and is preferably determined by considering a molecular diameter of an antibody and the like such that, when an antibody is immobilized, a part to be selectively digested is positioned near a surface layer of a pore.
  • An average pore diameter (D2) of a porous body is appropriately set, for example, in a range of about 10 nm-200 nm and in a range smaller than an average particle size (D1) of fine particles.
  • the average pore diameter (D2) of a porous body is preferably about 20 nm-200 nm, more preferably in ranges of 30 nm-150 nm, 40 nm-120 nm, and 50 nm-100 nm, and is particularly preferable about 100 nm.
  • a porous body in which a linker molecule that site-specifically interacts with an antibody is immobilized in a pore of the porous body is preferably used.
  • a linker molecule Protein A, Protein G or the like that site-specifically binds with an Fc domain of an antibody is preferably used.
  • an Fc domain of an antibody is immobilized in a pore, and an Fab domain is positioned near a surface layer of the pore. In this way, by controlling orientation of an antibody in a pore, regioselective digestion of an Fab domain, particularly, a CDR2, by a protease becomes possible.
  • a size of a linker molecule is selected such that a selective cleavage site of an antibody is positioned near a surface layer of a pore.
  • a molecular size in a state in which a linker molecule and an antibody are bound to each other is preferably about 0.5 times-1.5 times, more preferably about 0.6 times-1.2 times, even more preferably about 0.7 times-1.1 times, and particularly preferably about 0.8 times-1 times a pore diameter of a porous body.
  • a linker molecule is not fixed to a porous body and an antibody is directly bound in pores, it is preferable that a molecular diameter of the antibody and a pore diameter of the porous body satisfy the above relation.
  • a porous body that can be suitably used in the present invention is not particularly limited.
  • Protein G Ultralink resin manufactured by Pierce Corporation
  • Toyopearl manufactured by (TOSO Co. Ltd.)
  • the like can be used.
  • the Protein G Ultralink resin it is known that 95% of Protein G bound to resin is in pores.
  • a method for immobilizing an antibody in a pore of a porous body is not particularly limited. An appropriate method can be adopted according to characteristics of the antibody, the porous body or a linker molecule and the like. For example, when an antibody is immobilized in a porous body in which a protein A or a protein G is immobilized in a pore, by mixing a suspension of the porous body with a solution containing the antibody, the antibody can be easily immobilized in a pore.
  • a quantitative ratio of a porous body to an antibody can be appropriately set according to a purpose. For example, when a quantitative analysis of an antibody is performed, it is desirable that almost the entire antibody in a sample be immobilized in the porous body. Therefore, it is preferable that a quantitative ratio be set such that an amount of the porous body becomes excessive with respect to an estimated content of the antibody in the sample.
  • fine particles are used for a purpose of immobilizing a protease on surfaces of the fine particles and controlling access of the protease to an antibody immobilized in pores of a porous body. Therefore, the fine particles have a larger average particle size (D1) than the average pore diameter (D2) of the porous body so as not to enter deep into the pores of the porous body.
  • the fine particles are not particularly limited in shape. However, from a point of view of homogenization of access of the protease to the pores of the porous body, spherical fine particles are preferred. Further, it is preferable that the fine particles have a uniform particle size.
  • the average particle size (D1) of the fine particles is preferably in a range of 50 nm-500 nm, and is more preferably 1.2 or more times, even more preferably 1.5 or more times, and particularly preferably 1.8 or more times, for example, about 2 times the average pore size (D2) of the porous body.
  • the average particle size (D1) of the fine particles is preferably 100 nm or more, and more preferably 150 nm or more.
  • the average particle size of the fine particles is preferably 120 nm or more, more preferably 150 nm or more, and particularly preferably 170 nm or more.
  • an upper limit of the average particle size (D1) of the fine particles is preferably 500 nm or less, and more preferably 300 nm or less.
  • a material of the fine particles is not particularly limited as long as the above protease can be immobilized on surfaces of the fine particles.
  • a metal, a resin, or the like can be appropriately used as the material of the fine particles. Further, a metal coated with a resin, a resin coated with a metal, or the like can also be used.
  • magnetic fine particles that can be dispersed or suspended in an aqueous medium and can be easily recovered from the dispersion or suspension by application of a magnetic field are preferable. Further, from a point of view that aggregation is less likely to occur, magnetic fine particles coated with an organic polymer are more preferable.
  • base materials of magnetic fine particles include ferromagnetic alloys such as iron oxide (magnetite (Fe 3 O 4 ), maghemite ( ⁇ -Fe 2 O 3 )), and ferrite (Fe/M) 3 O 4 .
  • FG beads polymer magnetic fine particles having a particle size of about 200 nm obtained by coating ferrite particles with polyglycidyl methacrylate (poly GMA)) manufactured by Tamagawa Seiki Co., Ltd. is suitably used.
  • a spacer molecule having the above molecular diameter and capable of immobilizing a protease is preferably a non-protein, and is preferably a molecule having a functional group at a terminal, examples of the functional group including an amino group, a carboxyl group, an ester group, an epoxy group, a tosyl group, a hydroxyl group, a thiol group, an aldehyde group, a maleimide group, a succinimide group, an azide group, a biotin, an avidin, and a chelate.
  • a spacer molecule having an activated ester group is preferred.
  • a hydrophilic molecule can be used, examples of the hydrophilic molecule including polyethylene glycol and its derivatives, polypropylene glycol and its derivatives, polyacrylamide and its derivatives, polyethyleneimine and its derivatives, poly (ethylene oxide) and its derivatives, poly (ethylene terephthalic acid) and its derivatives, and the like.
  • Fine particles that are surface-modified with such spacer molecules are also commercially available, and those commercially available fine particles can be used.
  • fine particles modified with a spacer molecule having an ester group (active ester group) activated with N-hydroxysuccinimide is commercially available under a trade name “FG beads NHS” (Tamagawa Seiki Co., Ltd.).
  • the FG beads NHS has a particle size of about 200 nm ⁇ 20 nm, and is very homogeneous as fine particles.
  • a protease to be immobilized on fine particles a protease having trypsin activity and chymotrypsin activity is used.
  • a trypsin has high substrate specificity and has Lys or Arg at a C terminal of a peptide after cleavage and thus can homogenize a charge amount and charge localization of a peptide, and is suitable for preparation of a sample for mass spectrometry. Further, a trypsin has a small molecular diameter (about 3.8 nm) and an active site exists inside a molecule. Therefore, a region where the active site can access an antibody is restricted, and regioselectivity of protease digestion can be improved.
  • the present inventors have found that, by using a protease having trypsin activity and chymotrypsin activity, not a trypsin of a mass spectrometry grade that is usually used, peptide fragments optimal for quantitative detection of individual antibodies can be obtained.
  • a native trypsin can be used that is derived from a living body and is known to exhibit chymotrypsin-like activity by self-digestion.
  • a trypsin having improved resistance to self-digestion by subjecting a lysine residue of the trypsin to reductive methylation is commercially available as a trypsin of a mass spectrometry grade.
  • a trypsin used in the present invention is preferably a trypsin for which such a reductive dimethylation treatment of a lysine residue is not performed.
  • the method of the present invention allows a monoclonal antibody, which cannot be quantitatively detected when a trypsin that does not have chymotrypsin activity (for example, a trypsin of a mass spectrometry grade) is used, to be quantitatively detected using a peptide fragment that cannot be obtained by digestion of a trypsin of a mass spectrometry grade.
  • a trypsin that does not have chymotrypsin activity for example, a trypsin of a mass spectrometry grade
  • protease examples include Trypsin TPCK-Treated (manufactured by Sigma Corporation), and the like.
  • a method for immobilizing a protease on surfaces of fine particles is not particularly limited. An appropriate method can be adopted according to characteristics of the protease and the fine particles (or spacer molecules modifying the surfaces of the fine particles). For example, when the protease is immobilized on spacer-modified surfaces of the fine particles, by mixing a suspension of the fine particles and a solution containing the protease, the protease can be immobilized on the surfaces of the fine particles.
  • a method of amine coupling of the fine particles and the protease via functional groups of the spacer molecules is preferable.
  • a carboxyl group modifying surfaces of fine particles can be esterified with N-hydroxysuccinimide (NHS) to form an activated ester group to which an amino group of a protease can be bound.
  • NHS N-hydroxysuccinimide
  • This coupling reaction can be performed in the presence of carbodiimide as a condensing agent, examples of the carbodiimide including 1-ethyl-3-(3-dimethylaminopropyl) carbodiimide (EDAC), N,N′-dicyclohexylcarbodiimide (DCC), bis(2,6-diisopropylphenyl) carbodiimide (DIPC), and the like.
  • an amino group of a protease may be bound to an amino group modifying surfaces of fine particles using a cross-linking agent such as glutaraldehyde, bifunctional succinimide, bis(sulfosuccinimidyl) suberate (BS3), sulfonyl chloride, maleimide, and pyridyl disulfide.
  • a cross-linking agent such as glutaraldehyde, bifunctional succinimide, bis(sulfosuccinimidyl) suberate (BS3), sulfonyl chloride, maleimide, and pyridyl disulfide.
  • the coupling method of the fine particles and the protease via the functional groups of the spacer molecules can be performed by a simple operation of adding a protease solution to a suspension of the fine particles and mixing and stirring the mixture under certain conditions.
  • the protease is immobilized on the surfaces of the fine particles, it is preferable to inactivate an active portion that is not bound to the protease on the surfaces of the fine particles.
  • the antibody is protease-digested and peptide fragments are produced.
  • the contact preferably occurs in a liquid.
  • liquid means that a substrate (solid phase) and an enzyme (solid phase) are in contact with each other in a liquid phase, and is intended to mean an aqueous medium suitable for a protease digestion reaction.
  • Conditions of protease digestion in the present invention are not particularly limited, and conditions similar to general protease digestion can be suitably adopted.
  • a quantitative mixing ratio of the porous body on which the antibody is immobilized to the fine particles on the surfaces of which the protease is immobilized is not particularly limited, and may be set so as to have an amount of the protease corresponding to an amount of the antibody.
  • antibody:protease is preferably about 30:1-3:1, more preferably about 15:1-4:1, and even more preferably about 10:1-5:1.
  • protease digestion is performed in a state in which the antibody is immobilized on the porous body. Since the peptide fragments produced by the protease digestion exist in the liquid phase, target peptide fragments can be regioselectively obtained without performing an antibody elution or denaturation operation. According to the method of the present invention, it is possible to regioselectively collect a peptide fragment with a simple operation as compared to a conventional method.
  • a sample before being subjected to mass spectrometry is subjected to separation and concentration using liquid chromatography (LC).
  • LC liquid chromatography
  • an eluate from LC may be directly ionized and subjected to mass spectrometry.
  • Analysis can also be performed using LC/MS/MS or LC/MSn combining LC with tandem mass spectrometry.
  • the eluate from the LC may be collected once and then subjected to mass spectrometry.
  • An LC column is not particularly limited, and a hydrophobic column such as C30, C18, C8, and C4 generally used in peptide analysis, a carrier for hydrophilic affinity chromatography, and the like can be appropriately selected and used.
  • Mass spectrometry can determine an amino acid sequence and thus can determine whether or not a peptide fragment is a peptide fragment derived from a specific protein such as an antibody. Further, based on a peak intensity, concentration of peptide fragments in a sample can be determined. In performing analysis, a sample may be used for the analysis after being subjected to treatments such as desalting, solubilization, extraction, concentration, and drying when necessary.
  • An ionization method in mass spectrometry is not particularly limited, and an electron ionization (EI) method, a chemical ionization (CI) method, a field desorption (FD) method, a fast atom collision (FAB) method, a matrix assisted laser desorption ionization (MALDI) method, an electrospray ionization (ESI) method, and the like can be adopted.
  • EI electron ionization
  • CI chemical ionization
  • FD field desorption
  • FAB fast atom collision
  • MALDI matrix assisted laser desorption ionization
  • ESI electrospray ionization
  • a method for analyzing an ionized sample is also not particularly limited, and a method of a magnetic field deflection type, a quadrupole (Q) type, an ion trap (IT) type, a time of flight (TOF) type, a Fourier transform ion cyclotron resonance (FT-ICR) type, or the like can be appropriately determined according to the ionization method. Further, MS/MS analysis or multistage mass spectrometry of MS3 or higher can also be performed using a triple quadrupole mass spectrometer or the like.
  • a mass spectrometer that is suitable for implementing the method of the present invention is not particularly limited.
  • the mass spectrometer include liquid chromatography-triple quadrupole mass spectrometers such as LCMS-8030, LCMS-8040, and LCMS-8050 (all manufactured by Shimadzu Corporation), and mass spectrometers for structural analysis that performs precision mass analysis such as LCMS-IT-TOF and LCMS-Q-TOF (all manufactured by Shimadzu Corporation).
  • a target antibody When a peptide fragment containing an amino acid sequence of a CDR2 region specific to an antibody can be detected, a target antibody can be identified and quantified. Based on a mass spectrometry result, in order to identify an antibody, an existing database can also be used. Further, it is also possible to identify antibody by identifying an amino acid sequence of a peptide fragment using multistage mass spectrometry or the like.
  • the number of amino acid residues of a peptide to be detected is preferably about 5-30, and more preferably about 7-25.
  • an amount of the antibody can be calculated based on a peak area or a peak intensity of a detected peptide fragment ion (in the case of multistage MS, a fragment ion obtained by cleavage of a parent ion). For example, based on a correlation between a predetermined calibration curve and a peak area, or a correlation between a peak area derived from an internal standard added to a sample and a peak area derived from the sample, concentration of peptide fragments in the sample is calculated, and, based on the concentration of the peptide fragments, an amount and concentration of the antibody are calculated.
  • a peptide fragment to be detected in the method of the present invention has an amino acid sequence containing an amino acid derived from a CDR2 region of a heavy chain or a light chain of a monoclonal antibody of an antibody drug or the like.
  • Amino acid sequence information and the like of a monoclonal antibody intended to be used as an antibody drug are published, and information about amino acid sequences of a heavy chain and a light chain, Fab and Fc domains, CDR regions, a disulfide bond, and the like can be obtained. Further, according to the method of the present invention, contact between an antibody and a protease is limited, and thus, a type of a peptide obtained by selective protease digestion is also limited. Therefore, a person skilled in the art can predict an amino acid sequence of a peptide fragment digested by a specific protease.
  • detection and quantification of an antibody can also be performed by performing detection of multiple predicted peptide fragments concurrently in parallel.
  • an optimal peptide fragment is known, it is possible to determine a mass spectrometry condition suitable only for detection of the peptide fragment and it becomes possible to provide such information.
  • the method of the present invention includes, as one embodiment, selecting a peptide fragment containing an amino acid of a CDR2 region suitable for quantification of an individual monoclonal antibody. Further, another embodiment of the present invention provides a method that includes detecting a peptide fragment containing an amino acid of a CDR2 region selected as suitable for quantification of an individual monoclonal antibody.
  • peptides predicted to be suitable for mass spectrometry are peptides expressed by SEQ ID NO: 3-9.
  • concentration-dependent quantitative detection of these peptides could not be performed in serum samples, presumably due to interference of endogenous antibodies.
  • FSGVPDR SEQ ID NO: 4
  • FTISR SEQ ID NO: 5
  • FSGSGTDFTLK SEQ ID NO: 6
  • NSLYLQMNSLR SEQ ID NO: 7
  • HSDGNFAFGYWGQGTLVTVSSASTK SEQ ID NO: 8
  • GLEWVATISSASTYSYYPDSVK SEQ ID NO: 9
  • SEQ ID NO: 10 SYYPDSVK (SEQ ID NO: 11) GMSWVR (SEQ ID NO: 12) ISRVEAEDVGVY (SEQ ID NO: 13) YLQKPGQSPQLLIYK (SEQ ID NO: 14) NIVHINGDTYLEW
  • a monoclonal antibody as a measurement target is mogamulizumab
  • detection can be performed by multiple reaction monitoring using a triple quadrupole mass spectrometer (for example, LCMS-8050 manufactured by Shimadzu Corporation) with conditions described in Table 1 as indicators.
  • the present invention relates to a kit for quantitative detection of a monoclonal antibody using the above-described high-speed liquid chromatography-mass spectrometry (LC-MS), the kit including: a porous body for immobilizing a monoclonal antibody as a measurement target; fine particles onto which a protease having trypsin activity and chymotrypsin activity is immobilized; a reaction container for selectively digesting the monoclonal antibody by bringing the porous body into contact with the fine particles; and a buffer solution that is introduced into the reaction container together with the fine particles and the porous body and is for causing a digestion reaction by the protease to occur.
  • LC-MS liquid chromatography-mass spectrometry
  • Measurement using mass spectrometry enables very high-precision analysis.
  • proper sample preparation and setting of appropriate analysis conditions are very important.
  • the present invention provides a kit that can be used for implementing the above method.
  • the porous body and the fine particles included in the kit of the present invention are as described above.
  • the reaction container is not particularly limited as long as the reaction container is a container capable of allowing a monoclonal antibody immobilized on the porous body to be in contact with a protease immobilized on the fine particles in a liquid phase.
  • the reaction container is preferably a microtube or a plate.
  • reaction processes such as mixing using a vortex or a rotator performed for a reaction and filtration for separation of a peptide and the porous body and the fine particles after the reaction, a person skilled in the art can envision an appropriate reaction container.
  • the buffer solution included in the kit of the present invention is introduced into the reaction container together with the fine particles and the porous body and is for causing a digestion reaction by the protease to occur, and provides a reaction condition suitable for protease digestion.
  • Reaction conditions can be suitably determined depending on a protease and the like to be selected, and composition of the buffer solution can also be suitably determined.
  • the kit of the present invention can further include a filtration membrane for removing the porous body and the fine particles after the protease digestion reaction and extracting a product of the digestion reaction together with the buffer solution. This is because it is necessary to remove the porous body and the fine particles in order to subject a target peptide fragment obtained by the protease digestion to mass spectrometry.
  • the filtration membrane preferably functions as “a bottom of the reaction container” that substantially does not allow the buffer solution and peptides generated by protease digestion to pass through under a condition that a pressure or a centrifugal force is not applied, and functions as “a strainer” that allows the buffer solution and the peptides to pass through during an operation such as centrifugal separation.
  • a condition of centrifugal separation in order for the filtration membrane to allow the buffer solution and the peptides to pass through is not particularly limited, and, for example, a range of 3,000-10,000 g is preferable.
  • a filtration membrane made of polyvinylidene fluoride (PVDF) low-binding hydrophilic PVDF, pore diameter: 0.2 ⁇ m, manufactured by Millipore Corporation
  • PVDF polyvinylidene fluoride
  • kits for simultaneously processing multiple specimens
  • a kit can be used in which the filtration membrane is made of PVDF, and a housing material is a polyacrylonitrile resin, for example, Barex (registered trademark) (manufactured by Mitsui Fine Chemicals Inc.).
  • the kit of the present invention can further include an instruction manual describing use of the kit and/or conditions of mass spectrometry for detecting a monoclonal antibody.
  • the kit of the present invention can further include one or more internal standard peptides.
  • the internal standard peptides provide more reliable analysis results by being analyzed at the same time as specimens or separately under the same conditions as specimens.
  • the internal standard peptides contain a specific amino acid sequence of a monoclonal antibody as a measurement target, and are peptides generated by digestion by the protease included in the kit of the present invention.
  • the internal standard peptides may include those labeled with a stable isotope amino acid. In this case, since mass spectrometry quantification conditions are different as compared with peptides that do not contain isotopes, it is preferable to enclose quantification conditions for internal standards.
  • the internal standard peptides can include a peptide having an amino acid sequence of SEQ ID NO: 10.
  • kit of the present invention preparation operation of a peptide fragment for identification and quantification of a monoclonal antibody can be more conveniently performed, and automation using devices can also be easily achieved.
  • a protease is provided as a component of the kit in a state of being immobilized on surfaces of the fine particles, the preparation operation of the peptide fragment can be further simplified.
  • the present invention further provides a computer readable recording medium and a method package.
  • the recording medium is for use in the method of the present invention, and data for executing mass spectrometry is recorded in the recording medium, the data including data of a parent ion, a fragment ion, and a voltage applied to electrodes of a mass spectrometer, with respect to a peptide obtained by protease digestion of the monoclonal antibody.
  • the method package is for quantitative detection of a monoclonal antibody using liquid chromatography-mass spectrometry (LC-MS), and includes the recording medium.
  • LC-MS liquid chromatography-mass spectrometry
  • the term “method package” means independently distributable package that includes, in a readable form, an analysis condition of liquid chromatography-mass spectrometry with respect to a specific measurement target. By importing the data contained in the method package into LC-MS, it is possible to perform analysis with an optimum measurement condition obtained after detailed examination.
  • the method package can include an instruction manual of the recording medium.
  • the present invention provides a computer readable recording medium.
  • the recording medium is for use in a method for detecting a monoclonal antibody by analyzing a peptide fragment using liquid chromatography-mass spectrometry (LC-MS), the peptide fragment being obtained by performing selective protease digestion of the monoclonal antibody by bringing a porous body, in which the monoclonal antibody as a measurement target is immobilized in pores, into contact with fine particles, on which a protease having trypsin activity and chymotrypsin activity is immobilized.
  • LC-MS liquid chromatography-mass spectrometry
  • the data is not limited, but may include, for example, at least data of a parent ion, a fragment ion, and voltages applied to electrodes of a mass spectrometer, for example, triple quadrupoles (a first quadrupole, a second quadrupole, and a third quadrupole), with respect to a peptide that is obtained by protease digestion of the monoclonal antibody and has an amino acid sequence containing an amino acid of a CDR2 region.
  • the recording medium can further include data such as an expected retention time.
  • the expected retention time, the voltage data, and the like are numerical values that vary depending on equipment to be used and measurement conditions and the like, and are preferably provided in accordance with the equipment. Further, to facilitate understanding by a person skilled in the art, for a numerical value that varies depending on a condition, it is preferable to also provide a variation range of the numerical value.
  • the recording medium may be of any form, and is not particularly limited. Examples of the recording medium include disks and memories capable of magnetically or optically recording information.
  • the above method package can include, for example, the following information and software functions.
  • the present invention also provides a method package for detection of a monoclonal antibody using liquid chromatography-mass spectrometry (LC-MS), the method package including the above-described recording medium.
  • the method package can include an instruction manual of the recording medium.
  • An example of the above-described recording medium or method package is one that includes only information specific to a specific monoclonal antibody. Therefore, when the monoclonal antibody is, for example, mogamulizumab, it is possible to provide a recording medium or a method package in which conditions suitable for analyzing mogamulizumab are described.
  • the data included in the recording medium can be related to, for example, analysis conditions with respect to a peptide having an amino acid sequence of SEQ ID NO: 10.
  • the method package may describe data common to multiple mass spectrometers or may describe various data suitable for analysis using a specific mass spectrometer
  • the recording medium or method package can be provided together with the above-described kit of the present invention above or separately from the kit.
  • a protease was immobilized on fine particles by the following steps.
  • Trypsin Gold 1 mg (manufactured by Promega Corporation) or Trypsin TPCK 5 mg (manufactured by Sigma-Aldrich Corporation) is opened and dissolved in 25 mM HEPES-NaOH at pH 7.0 cooled to 4° C. After the dissolution, the solution is put in a 50 ml centrifuge tube, which is then placed on ice. The enzyme is washed once with 25 mM HEPES-NaOH at pH 7.0 and is collected as much as possible. A final buffer volume is about 25 ml.
  • a Trypsin Gold or Trypsin TPCK solution is added to 200 mg of a precipitate of FG beads. After a suspension of FG beads is confirmed using an ice-cooled ultrasonic washer, vortex is continuously performed for 30 minutes at a minimum speed that allows the suspension to be maintained.
  • centrifugal separation (15,000 g ⁇ 5 minutes, 4° C.) is performed, and supernatant is removed. The supernatant is collected and a coupling efficiency confirmation test based on a BCA assay is performed.
  • Tris-HCl at pH 8.0 25 mM Tris-HCl at pH 8.0 is added such that a protease final concentration is 0.5 ⁇ g/ ⁇ l.
  • the solution is dispensed to 500 ⁇ l. Thereafter, the solution is stored at ⁇ 80° C.
  • Protease digestion of a monoclonal antibody in a plasma sample was performed by the following steps.
  • 20 ⁇ l of a plasma is diluted with 180 ⁇ l of PBS+0.1% n-octyl- ⁇ -D-thioglucoside (manufactured by Dojin Chemical).
  • An antibody drug was spiked into a commercially available plasma (manufactured by Sigma Corporation) and a peptide obtained by protease digestion was detected.
  • mogamulizumab drug name: POTELIGEO, Kyowa Hakko Kirin Co., Ltd.
  • Conditions of mass spectrometry are as follows.
  • Solvent A 0.1% formic acid
  • solvent B 0.1% formic acid+acetonitrile
  • Heat block temperature 400° C.
  • FIG. 1 illustrates amino acid sequences (SEQ ID NO: 1 and 2) of a heavy chain and a light chain of mogamulizumab, and sequences (SEQ ID NO: 3-9) of cleavage fragments due to Trypsin Gold used for quantification and positions of the sequences (SEQ ID NO: 3-9) in the amino acid sequence of the heavy chain or the light chain.
  • FIG. 2 illustrates results of subjecting the Trypsin Gold cleavage fragments (SEQ ID NO: 3-9) of mogamulizumab to MRM chromatogram according to the above-described analysis conditions.
  • SEQ ID NO: 3-9 Trypsin Gold cleavage fragments
  • FIG. 3 illustrates results of a preparing calibration curves based on peaks detected with Trypsin Gold cleavage fragments (SEQ ID NO: 3-9) spiked into a plasma.
  • Trypsin Gold is used as a protease, it was not possible to prepare calibration curves according to an amount of peptide fragments spiked into a plasma, and it was found that none of these peptide fragments is suitable for concentration-dependent quantitative detection.
  • FIG. 4 illustrates sequences (SEQ ID NO: 10-14) of Trypsin TPCK cleavage fragments used in quantification of mogamulizumab, and positions of the sequences in the amino acid sequences of the heavy chain and the light chain.
  • FIG. 5 illustrates results of analyzing the Trypsin TPCK cleavage fragments (SEQ ID NO: 10-14) of mogamulizumab under the same conditions as above. From the results of FIG. 5 , in both the case where mogamulizumab raw powder was directly analyzed (upper figure) and the case where mogamulizumab was spiked into a plasma (lower figure), peaks derived from five kinds of peptide fragments were confirmed.
  • the peptide SYYPDSVK (SEQ ID NO: 10) was selected as a mogamulizumab target peptide in precision mass spectrometry and structure identification by precision mass spectrometry was performed, and the results are shown in FIG. 7 . From the results of FIG. 7 , it was confirmed that the structure as predicted from the peptide sequence can be identified.
  • the detected peptide SYYPDSVK (SEQ ID NO: 10) is derived from an Fv region, and is positioned on an outermost side in a three-dimensional structure of a human immunoglobulin molecule obtained in Protein Data Bank.
  • an antibody in a sample can be quantified by selectively digesting the antibody using a specific protease and subjecting a resulting peptide fragment to mass spectrometry.
  • the method of the present invention allows easy operation and can ensure reproducibility and quantitativeness.
  • the present invention can provide an extremely versatile analysis method for such a wide variety of antibody drugs, and can contribute to acceleration of future antibody drug development.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Biochemistry (AREA)
  • Wood Science & Technology (AREA)
  • Zoology (AREA)
  • Immunology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Physics & Mathematics (AREA)
  • Analytical Chemistry (AREA)
  • General Engineering & Computer Science (AREA)
  • Genetics & Genomics (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • General Physics & Mathematics (AREA)
  • Pathology (AREA)
  • Biotechnology (AREA)
  • Microbiology (AREA)
  • Biomedical Technology (AREA)
  • Hematology (AREA)
  • Urology & Nephrology (AREA)
  • Biophysics (AREA)
  • Electrochemistry (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Cell Biology (AREA)
  • Food Science & Technology (AREA)
  • Medicinal Chemistry (AREA)
  • Other Investigation Or Analysis Of Materials By Electrical Means (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
  • Enzymes And Modification Thereof (AREA)
US15/578,374 2015-06-01 2015-06-01 Method for quantifying monoclonal antibody Abandoned US20180148763A1 (en)

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
PCT/JP2015/065806 WO2016194114A1 (ja) 2015-06-01 2015-06-01 モノクローナル抗体の定量方法

Publications (1)

Publication Number Publication Date
US20180148763A1 true US20180148763A1 (en) 2018-05-31

Family

ID=57440329

Family Applications (1)

Application Number Title Priority Date Filing Date
US15/578,374 Abandoned US20180148763A1 (en) 2015-06-01 2015-06-01 Method for quantifying monoclonal antibody

Country Status (8)

Country Link
US (1) US20180148763A1 (de)
EP (1) EP3306310B1 (de)
JP (1) JP6537605B2 (de)
CN (1) CN107709980B (de)
CA (1) CA2987005C (de)
SG (1) SG11201709908QA (de)
TW (1) TWI675917B (de)
WO (1) WO2016194114A1 (de)

Cited By (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10704021B2 (en) 2012-03-15 2020-07-07 Flodesign Sonics, Inc. Acoustic perfusion devices
US10724029B2 (en) 2012-03-15 2020-07-28 Flodesign Sonics, Inc. Acoustophoretic separation technology using multi-dimensional standing waves
US10785574B2 (en) 2017-12-14 2020-09-22 Flodesign Sonics, Inc. Acoustic transducer driver and controller
US10975368B2 (en) 2014-01-08 2021-04-13 Flodesign Sonics, Inc. Acoustophoresis device with dual acoustophoretic chamber
US11085035B2 (en) 2016-05-03 2021-08-10 Flodesign Sonics, Inc. Therapeutic cell washing, concentration, and separation utilizing acoustophoresis
US11209392B2 (en) 2017-12-28 2021-12-28 Shimadzu Corporation Simplified monoclonal antibody quantification method
US11214789B2 (en) 2016-05-03 2022-01-04 Flodesign Sonics, Inc. Concentration and washing of particles with acoustics
US11377651B2 (en) 2016-10-19 2022-07-05 Flodesign Sonics, Inc. Cell therapy processes utilizing acoustophoresis
US11459540B2 (en) 2015-07-28 2022-10-04 Flodesign Sonics, Inc. Expanded bed affinity selection
US11474085B2 (en) 2015-07-28 2022-10-18 Flodesign Sonics, Inc. Expanded bed affinity selection
US11708572B2 (en) 2015-04-29 2023-07-25 Flodesign Sonics, Inc. Acoustic cell separation techniques and processes

Families Citing this family (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10967298B2 (en) 2012-03-15 2021-04-06 Flodesign Sonics, Inc. Driver and control for variable impedence load
US9950282B2 (en) 2012-03-15 2018-04-24 Flodesign Sonics, Inc. Electronic configuration and control for acoustic standing wave generation
US11021699B2 (en) 2015-04-29 2021-06-01 FioDesign Sonics, Inc. Separation using angled acoustic waves
JP2020513248A (ja) 2016-10-19 2020-05-14 フロデザイン ソニックス, インク.Flodesign Sonics, Inc. 音響による親和性細胞抽出
AU2019264349A1 (en) * 2018-04-30 2020-10-15 Phynexus Inc. Qualitative analysis of proteins
CN108680637A (zh) * 2018-05-15 2018-10-19 苏州大学 基于马来酰亚胺衍生物组合的非同位素双标记方法
JPWO2022270126A1 (de) 2021-06-23 2022-12-29

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060014212A1 (en) * 2002-05-10 2006-01-19 Epitome Biosystems, Inc. Proteome epitope tags and methods of use thereof in protein modification analysis
US20160252522A1 (en) * 2013-09-09 2016-09-01 Shimadzu Corporation Method for preparing peptide fragments, kit for preparing peptide fragments to be used therein, and analysis method

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10151759B2 (en) * 2011-02-10 2018-12-11 Hoffmann-La Roche Inc. Method for the purification of a glycan and/or a glycoconjugate by chromatography using a stationary phase comprising cotton

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060014212A1 (en) * 2002-05-10 2006-01-19 Epitome Biosystems, Inc. Proteome epitope tags and methods of use thereof in protein modification analysis
US20160252522A1 (en) * 2013-09-09 2016-09-01 Shimadzu Corporation Method for preparing peptide fragments, kit for preparing peptide fragments to be used therein, and analysis method

Cited By (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10704021B2 (en) 2012-03-15 2020-07-07 Flodesign Sonics, Inc. Acoustic perfusion devices
US10724029B2 (en) 2012-03-15 2020-07-28 Flodesign Sonics, Inc. Acoustophoretic separation technology using multi-dimensional standing waves
US10975368B2 (en) 2014-01-08 2021-04-13 Flodesign Sonics, Inc. Acoustophoresis device with dual acoustophoretic chamber
US11708572B2 (en) 2015-04-29 2023-07-25 Flodesign Sonics, Inc. Acoustic cell separation techniques and processes
US11459540B2 (en) 2015-07-28 2022-10-04 Flodesign Sonics, Inc. Expanded bed affinity selection
US11474085B2 (en) 2015-07-28 2022-10-18 Flodesign Sonics, Inc. Expanded bed affinity selection
US11085035B2 (en) 2016-05-03 2021-08-10 Flodesign Sonics, Inc. Therapeutic cell washing, concentration, and separation utilizing acoustophoresis
US11214789B2 (en) 2016-05-03 2022-01-04 Flodesign Sonics, Inc. Concentration and washing of particles with acoustics
US11377651B2 (en) 2016-10-19 2022-07-05 Flodesign Sonics, Inc. Cell therapy processes utilizing acoustophoresis
US10785574B2 (en) 2017-12-14 2020-09-22 Flodesign Sonics, Inc. Acoustic transducer driver and controller
US11209392B2 (en) 2017-12-28 2021-12-28 Shimadzu Corporation Simplified monoclonal antibody quantification method

Also Published As

Publication number Publication date
CA2987005A1 (en) 2016-12-08
WO2016194114A1 (ja) 2016-12-08
JP6537605B2 (ja) 2019-07-03
CN107709980A (zh) 2018-02-16
TWI675917B (zh) 2019-11-01
EP3306310B1 (de) 2020-08-05
JPWO2016194114A1 (ja) 2018-03-22
CA2987005C (en) 2021-11-16
EP3306310A1 (de) 2018-04-11
EP3306310A4 (de) 2018-11-21
TW201643252A (zh) 2016-12-16
SG11201709908QA (en) 2017-12-28
CN107709980B (zh) 2020-11-06

Similar Documents

Publication Publication Date Title
CA2987005C (en) Method for quantifying monoclonal antibody
EP3270154B1 (de) Verfahren zur detektion eines monoklonalen antikörpers unter verwendung von massenspektrometrie
US11619616B2 (en) Method for quantifying monoclonal antibody having antigen or anti-antibody bonded thereto
US20220137062A1 (en) Kit for preparing sample for detecting monoclonal antibody
WO2018167847A1 (ja) モノクローナル抗体の同時定量方法
KR101826449B1 (ko) 펩티드 단편의 조제 방법 및 그것에 사용되는 펩티드 단편 조제용 킷, 및 분석 방법
US20200309735A1 (en) Simplified Monoclonal Antibody Quantification Method
US20180051053A1 (en) Method for obtaining peptide fragment from antibody by protease decomposition reaction with restricted reaction field
WO2022270126A1 (ja) 抗体を分析する方法

Legal Events

Date Code Title Description
AS Assignment

Owner name: NATIONAL CANCER CENTER, JAPAN

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:SHIMADA, TAKASHI;IWAMOTO, NORIKO;HAMADA, AKINOBU;SIGNING DATES FROM 20171023 TO 20171114;REEL/FRAME:044261/0443

Owner name: SHIMADZU CORPORATION, JAPAN

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:SHIMADA, TAKASHI;IWAMOTO, NORIKO;HAMADA, AKINOBU;SIGNING DATES FROM 20171023 TO 20171114;REEL/FRAME:044261/0443

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION